You are on page 1of 12

Neuropharmacology 148 (2019) 320e331

Contents lists available at ScienceDirect

Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm

N-Oleoyl-glycine reduces nicotine reward and withdrawal in mice


Giulia Donvito a, 1, Fabiana Piscitelli b, 1, Pretal Muldoon a, Asti Jackson a,
Rosa Maria Vitale b, Enrico D'Aniello b, f, Catia Giordano c,
Bogna M. Ignatowska-Jankowska a, Mohammed A. Mustafa a, Francesca Guida c,
Gavin N. Petrie d, Linda Parker d, Reem Smoum e, Laura Sim-Selley a, Sabatino Maione c,
Aron H. Lichtman a, g, *, M. Imad Damaj a, **, Vincenzo Di Marzo b, ***,
Raphael Mechoulam e
a
Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
b
Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Naples, Italy
c
Endocannabinoid Research Group, Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
d
Department of Psychology and Collaborative Neuroscience Graduate Program, University of Guelph, Guelph, ON, Canada
e
Institute for Drug Research, Medical Faculty, Hebrew University, Jerusalem, Israel
f
Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy
g
Department of Medicinal Chemistry, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA

a r t i c l e i n f o a b s t r a c t

Article history: Cigarette smokers with brain damage involving the insular cortex display cessation of tobacco smoking,
Received 6 September 2017 suggesting that this region may contribute to nicotine addiction. In the present study, we speculated that
Received in revised form molecules in the insular cortex that are sensitive to experimental traumatic brain injury (TBI) in mice
1 March 2018
might provide leads to ameliorate nicotine addiction. Using targeted lipidomics, we found that TBI eli-
Accepted 17 March 2018
cited substantial increases of a largely uncharacterized lipid, N-acyl-glycine, N-oleoyl-glycine (OlGly), in
Available online 19 March 2018
the insular cortex of mice. We then evaluated whether intraperitoneal administration of OlGly would
alter withdrawal responses in nicotine-dependent mice as well as the rewarding effects of nicotine, as
Keywords:
N-oleoyl glycine
assessed in the conditioned place preference paradigm (CPP). Systemic administration of OlGly reduced
Nicotine withdrawal mecamylamine-precipitated withdrawal responses in nicotine-dependent mice and prevented nicotine
Cannabinoid receptor-1 (CB1) CPP. However, OlGly did not affect morphine CPP, demonstrating a degree of selectivity. Our respective
Conditioned place preference (CPP) in vitro and in vivo observations that OlGly activated peroxisome proliferator-activated receptor alpha
Insular cortex (PPAR-a) and the PPAR-a antagonist GW6471 prevented the OlGly-induced reduction of nicotine CPP in
Peroxisome proliferator-activated receptor mice suggests that this lipid acts as a functional PPAR-a agonist to attenuate nicotine reward. These
alpha (PPAR-a) findings raise the possibility that the long chain fatty acid amide OlGly may possess efficacy in treating
nicotine addiction.
© 2018 Published by Elsevier Ltd.

1. Introduction

* Corresponding author. Department of Pharmacology and Toxicology, Virginia Dependence to tobacco represents one of the most frequent
Commonwealth University, PO Box 980613, Kontos Medical Sciences Building, 1217 causes of mortality and morbidity in the world (Doll et al., 2004).
East Marshall Street, Richmond, VA, 23298, USA. Nicotine, a primary constituent of tobacco responsible for its
** Corresponding author. Department of Pharmacology and Toxicology, Virginia
rewarding effects, acts on nicotinic acetylcholine receptors
Commonwealth University, PO Box 980613, Kontos Medical Sciences Building, 1217
East Marshall Street, Richmond, VA, 23298, USA.
(nAChRs), which are expressed on pre- and post-synaptic terminals
*** Corresponding author. Endocannabinoid Research Group, Institute of Bio- (Albuquerque et al., 2009) in a variety of CNS pathways, including
molecular Chemistry, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, the mesolimbic reward system (Watkins et al., 2000). Chronic to-
Comprensorio Olivetti, 80078, Pozzuoli, Napoli, Italy. bacco use often induces dependence and cessation can lead to af-
E-mail addresses: alichtma@vcu.edu (A.H. Lichtman), m.damaj@vcuhealth.org
fective (e.g., anxiety, anhedonia, depression, dysphoria,
(M.I. Damaj), vdimarzo@icb.cnr.it (V. Di Marzo).
1
These authors contributed equally to this work. hyperalgesia, and irritability), somatic (e.g., tremors, bradycardia,

https://doi.org/10.1016/j.neuropharm.2018.03.020
0028-3908/© 2018 Published by Elsevier Ltd.
G. Donvito et al. / Neuropharmacology 148 (2019) 320e331 321

gastrointestinal discomfort, and increased appetite), and cognitive water were available ad libitum. Male C57BL/6 mice (Charles River,
(e.g., difficulty concentrating and impaired memory) withdrawal Italy) weighing 18e20 g were used for the mild TBI Weight Drop
symptoms (Heishman et al., 2010). Naqvi and co-workers (Naqvi (WD) model.
et al., 2007) reported that smokers with brain damage involving All animal protocols were approved by the Virginia Common-
the insula, a region implicated in conscious urges, were more likely wealth University Institutional Animal Care and Use Committee,
than smokers with brain damage not involving the insula to un- were in accordance with the National Institutes of Health Guide for
dergo a disruption of smoking addiction. Although the neurobio- the Care and Use of Laboratory Animals and with the National In-
logical mechanisms underlying this association remain to be stitutes of Health Guide for the Care and Use of Laboratory Animals,
determined, preclinical data also implicate a role of the insular and by the Animal Ethics Committee of The Second University of
cortex in nicotine reward. Specifically, rats given long access to Naples, in compliance with Italian (D.L. 116/92) and European
nicotine self-administration had increased levels of dopamine and Commission (O.J. of E.C. L358/1 18/12/86) regulations on the pro-
cAMP-regulated phosphoprotein of 32 kD phosphorylated at the tection of laboratory animals.
protein kinase A site in the insular cortex (Abdolahi et al., 2010).
This observation along with human data (Naqvi et al., 2014) suggest 2.2. Synthesis of OlGly
a role of this neuroanatomical region in addictive behavior and that
neurochemicals within the insular cortex that are sensitive to brain To a solution of oleic acid (1 gm, 3.54 mmol) and N,N-dime-
injury may modulate nicotine addiction. thylformamide (266 mL, 3.64 mmol) in dry methylene chloride
As a starting point to test molecules that may be useful to treat (10 mL) was added dropwise oxalyl chloride (2.0 M solution in
nicotine addiction, we sought to identify neurochemicals in the methylene chloride, 3.5 mL, 7 mmol) under nitrogen atmosphere.
insular cortex of mice that may be altered by traumatic brain injury The reaction mixture was stirred for 1 h and then the solvent was
(TBI). Twenty-four h after TBI, we harvested insular cortex as well evaporated under a nitrogen flow. The crude material in methylene
as comparison regions (i.e., hippocampus, and hypothalamus) for chloride (10 mL) was added to a solution of glycine (800 mg,
analysis. We initially focused on endocannabinoids, because of 10.62 mmol) and 2 N potassium hydroxide in an ice bath. Then, the
their well-known involvement in nicotine reward and withdrawal reaction mixture was stirred for 1 h, water (10 mL) was added, and
signs in nicotine-dependent laboratory animals (Castan ~e
 et al., the mixture was acidified to pH 3 with 1 N HCl. The product was
2005, 2002; Gamaleddin et al., 2015; Le Foll and Goldberg, 2004; extracted with ether (3  50 mL) and dried (MgSO4), and solvent
Merritt et al., 2008; Valjent et al., 2002), as well as related lipid was evaporated under reduced pressure.
signaling molecules. Among several investigated lipids, in addition The crude material was chromatographed on silica gel (eluting
to the endocannabinoids N-arachidonoylethanolamine (ananda- with chloroform: methanol) to yield a crystalline solid. Melting
mide; AEA) (Devane et al., 1992) and 2-arachidonylglycerol (2-AG) point 93e94 C (degradation); LC-MS: (M-H)þ ¼ 339 m/z; NMR
(Mechoulam et al., 1995; Sugiura et al., 1995), we examined N- (CD3OH, ppm): 5.35e5.32 (m, 2H), 4.45 (s, 2H), 2.13e2.18 (m, 6H),
acylethanolamines, N-acyldopamines, N-acylserines and N- 1.58 (m, 2H), 1.32e1.29 (m, 20H), 0.88 (t, 3H).
acylglycines.
Based on the results of the lipidomic analyses of TBI and control 2.3. Drugs
brain areas, we selected a largely uncharacterized member of the
latter class, N-oleoyl glycine (OlGly) (Bradshaw et al., 2009). Spe- [2H]8AEA, [2H]52-AG, [2H]4 PEA, [2H]4 OEA, [2H]8 N-arach-
cifically, we tested whether exogenous administration of this idonoyldopamine (NADA), [2H]8AraSer and [2H]8AraGly were pur-
molecule would attenuate precipitated withdrawal responses in chased from Cayman Chemicals (MI, USA). OlGly was synthetized in
nicotine-dependent mice (Jackson et al., 2008). Additionally, we the Mechoulam laboratory. The PPAR-a receptor antagonist
used the conditioned place preference paradigm (CPP) to infer GW6471 [N-((2S)-2-(((1Z)-1-methyl-3-oxo-3-(4-(trifluoromethyl)
whether it would attenuate the rewarding effects of nicotine (Kota phenyl)prop-1-enyl)amino)-3-(4-(2-(5-methyl2phenyl-1,3-oxazol-
et al., 2007). In order to elucidate potential underlying mechanisms 4-yl) ethoxy)phenyl)propyl)propanamide] was purchased from
of action of OlGly, we examined two likely targets. First, we tested tocris (Minneapolis, MN).
whether it interacts with the endocannabinoid system. Specifically, CP55,940 (()-cis-3-[2-hydroxy-4-(1,1-dimethylheptyl)
we investigated if it binds to human recombinant CB1 and CB2 re- phenyl]-trans-4-(3-hydroxypropyl)-cyclohexanol) and morphine
ceptors transfected on HEK-293 cells, inhibits the primary AEA sulfate were generously provided by NIDA (Rockville, MD). OlGly,
hydrolytic enzyme fatty acid amide hydrolase (FAAH), or elicits CP55,940 and GW6471 were dissolved in a vehicle solution con-
in vivo pharmacological effects, as assessed in the tetrad assay, sisting of ethanol (5% of total volume), alkamuls-620 (Sanofi-
which is highly sensitive to CB1 receptor agonists (Little et al., 1988; Aventis, Bridgewater, NJ) (5% of total volume), and saline (0.9%
Martin et al., 1991). Second, we examined whether it binds and NaCl) (90% of total volume). OlGly and CP55,940 were given via the
produces its pharmacological effects through peroxisome intraperitoneal (i.p.) route of administration in a volume of 10 ml/
proliferator-activated receptor a (PPAR-a), which has been kg ()-Nicotine hydrogen tartrate [()-1-methyl-2-(3-pyridyl)
demonstrated to play a role in nicotine dependence (Jackson et al., pyrrolidine (þ)-bitartrate] and mecamylamine HCl were purchased
2017; Mascia et al., 2011; Panlilio et al., 2012). from Sigma-Aldrich Inc. (St. Louis, MO, USA). Morphine sulfate
[morphine hemi[sulfate pentahydrate]] Nicotine and mecamyl-
2. Material and methods amine (2 mg/kg) were dissolved in physiological saline and given
via the subcutaneous (s.c.) route of administration in a volume of
2.1. Subjects 10 ml/kg. For the nicotine CPP study, 0.5 mg/kg nicotine dose was
used because this dose reliably produces significant CPP in ICR
Male ICR mice (6e8 weeks old; Harlan, Indianapolis, IN) with a mouse (Kota et al., 2007). Morphine CPP was performed with
body mass of 27e32 g served as subjects in all in vivo pharmacology 10 mg/kg (s.c.) as recently described (Alajaji et al., 2016). For nico-
experiments. Mice were group-housed (four per cage) for at least tine withdrawal studies, 24 mg/kg/day nicotine or saline was
one week before the beginning of experiments on a 12/12 light/ continuously perfused for 14 days using s.c. Osmotic minipumps
dark cycle (lights on at 0600 h), with an ambient temperature of (model 2002; Alzet Corporation, Cupertino, CA) that were
20e22  C and humidity of 55e60%. Standard rodent chow and tap implanted under isoflurane anesthesia. This prolonged nicotine
322 G. Donvito et al. / Neuropharmacology 148 (2019) 320e331

administration regimen reliably produces significant withdrawal mobile phase with a flow rate of 0.15 ml/min. Identification of N-
syndrome in the three behavioral paradigms used here (Jackson acyldopamines, N-acylglycines and N-acylserines was carried out
et al., 2009). using ESI ionization in the positive mode with nebulizing gas flow
of 1.5 ml/min and curved desolvation line temperature of 250  C.
2.4. Surgical preparation and brain injury (mouse WD model)
2.6. Nicotine precipitated withdrawal studies
Experimental mild TBI (TBI) was performed using a weight-drop
device developed in the Naples laboratory. Mice were anesthetized The minipumps were subcutaneously implanted in mice under
with intraperitoneal injection of 250 mg/kg Avertin before being isoflurane anesthesia. The pumps delivered 24 mg/kg/day nicotine
subjected to TBI. After a midline longitudinal incision, the skull was or saline for 14 days. On the morning of day 15 (defined as 0 min),
exposed to locate the area of impact and placed under a metal tube all mice that received nicotine were given an injection of OlGly (10,
device where the opening was positioned directly over the animal's 30, or 60 mg/kg, i.p.) or vehicle (time: 0 min). At 15 min, each
head. The injury was induced by dropping a cylindrical metal mouse was given an s.c. injection of the non-selective nicotinic
weight (50 g), through a vertical metal guide tube from a height of acetylcholine receptor (nAChR) antagonist mecamylamine (2 mg/
20 cm. The point of impact was between the anterior coronal suture kg, s.c.). At 25 min, an experimenter, blinded to drug treatment,
(bregma) and posterior coronal suture (lambda). Immediately evaluated the mice in the elevated plus maze and then for physical
following injury, the skin was closed with surgical wound clips and (somatic) nicotine withdrawal signs, as previously described
mice were placed back in their cages to allow for recovery from the (Jackson et al., 2008). The mice were first evaluated for 5 min in the
anesthesia and TBI. Sham mice were submitted to the same pro- plus maze test in which the duration of time spent on the open
cedure as described for TBI, but without release of the weight. arms versus closed arms of the plus maze was assessed. The
number of arm crosses between the open and closed arms was also
2.5. Extraction and quantification of endocannabinoids, N- counted as a measure of locomotor activity. Immediately following
acylethanolamines, N-acyldopamines, N-acylserines and N- plus maze assessment, each mouse was placed in a clear activity
acylglycines cage without bedding for a 20 min observation period of somatic
signs measured that included paw and body tremors, head shakes,
Brain tissues were rapidly frozen, dounce-homogenized, and backing, jumps, curls, and ptosis. The total number of somatic signs
extracted with chloroform/methanol/Tris-HCl 50 mM pH 7.5 (2:1:1, was tallied for each mouse and the average number of somatic signs
v/v) containing internal deuterated standards for AEA, 2-AG, PEA, during the observation period was plotted for each test group. This
OEA, NADA, AraSer and AraGly quantification by isotope dilution testing sequence was chosen based on our prior studies showing
(5 pmol for [2H]8AEA; 50 pmol for [2H]52-AG, [2H]4 PEA and [2H]4 that this order of testing reduced within-group variability and
OEA; 10 pmol for [2H]8 NADA, [2H]8AraSer and [2H]8AraGly). Then produced consistent results (Jackson et al., 2008). An observer
the lipid extract was purified by open bed chromatography on sil- blinded to experimental treatment performed all studies.
ica. Fractions were eluted within increasing amounts of CH3OH in In order to test whether OlGly is brain penetrant, ICR mice
CHCl3 and part of the 9:1 (v/v) fraction was analyzed by liquid received a single administration of OlGly (60 mg/kg, i.p.), or vehicle.
chromatography-atmospheric pressure chemical ionization-single After 15 min, plasma, insular cortex, hippocampus and hypothala-
quadrupole mass spectrometry for AEA, 2-AG, PEA and OEA mus were harvested and processed for the extraction and quanti-
levels, as previously described (Bisogno et al., 2009; Piscitelli et al., fication of the levels of OlGl,y as described above. The dose of OlGly
2011). AEA, 2-AG, PEA and OEA levels were calculated on the basis used in this experiment and the time point for tissue collection
of their area ratio with the internal deuterated standard signal were based on the behavioral experiments.
areas. Part of the 9:1 fraction was used for N-acyldopamine iden-
tification, whereas the 7:3 fraction was used for N-acylglycine and 2.7. Conditioned place preference (CPP) studies
N-acylserine identification and quantification by LC-MS-IT-TOF
(Shimadzu Corporation, Kyoto, Japan) equipped with an ESI inter- An unbiased CPP paradigm was performed, as previously
face, using multiple reaction monitoring (MRM). The method for described (Kota et al., 2007). Briefly, the CPP apparatus consisted of
NADA was as previously described (Bisogno et al., 2009). Quanti- three chambers in a linear arrangement (MedAssociates, St. Albans,
fication was performed by isotope dilution by using m/z values of VT, ENV3013) with white and black chambers (20  20  20 cm
370.3192 and 362.2692 corresponding to the molecular ion each), which also differed in floor texture (white mesh or black
[MþH]þ for deuterated and undeuterated AraGly; or m/z values of rod). These chambers were separated by a small gray chamber with
400.3297 and 392.2795 corresponding to the molecular ion a smooth PVC floor. Partitions could be removed to allow access
[MþH]þ for deuterated and undeuterated AraSer The recovery of from the gray chamber to the black and white chambers. On day 1,
AraGly and AraSer from rat brain tissues using the extraction and animals were confined to the middle chamber for a 5-min habit-
analytical procedure reported here (see Methods) was 49.1 ± 15.7% uation period and then allowed to move freely among all three
and 42.1 ± 15.9% (n ¼ 7). The LC-ESI-IT-ToF method was specific and chambers for 15 min. Time spent in each chamber was recorded,
exhibited a limit of detection (LOD, defined as the concentration at and no systematic bias was observed in baseline chamber prefer-
which the signal/noise ratio is greater than 3:1) of 50 fmol in the ence. Twenty-min conditioning sessions occurred twice a day (days
MS mode, and 1 pmol in the MS/MS mode for all the compounds 2e4). During conditioning sessions, mice were confined to one of
analyzed. Moreover, the ratio between the [MþH]þ peak areas of the larger chambers. The control group received saline in one large
undeuterated (0.025e10 pmol) vs. deuterated (1 pmol) AraGly and chamber in the morning and saline in the other large chamber in
AraSer varied linearly with the amount of the respective deuterated the afternoon. The nicotine group received nicotine in one large
standards. The quantification limit of compounds was 100 fmol and chamber and saline in the other large chamber. Treatments were
the reproducibility of the method was 95%e99%. The chromato- counterbalanced equally in order to ensure that some mice
grams of the high-resolution [M þ H]þ values were extracted and received nicotine in the morning while others received it in the
used for calibration and quantification. LC analysis was performed afternoon. The nicotine-paired chamber was randomized among
in the isocratic mode using a Kinetex C18 Column (10 cm  2.1 mm, subjects. Sessions were 4 h apart and were conducted by the same
5 mm) and CH3OH/water/acetic acid (85:15:0.1 by vol.) as the investigator. On each of the conditioning days, mice were
G. Donvito et al. / Neuropharmacology 148 (2019) 320e331 323

pretreated with OlGly (i.p.) or vehicle 15 min prior to nicotine or as the high affinity ligand and displaced with 10 mM WIN 55212-2
morphine (s.c.) injection. Five min after nicotine administration as the heterologous competitor for nonspecific binding (Ki values:
(0.5 mg/kg, s.c.), subjects were given 20-min conditioning sessions. 9.2 nM and 2.1 nM respectively for CB1 and CB2 receptor). OlGly was
In the morphine CPP comparison study, mice were given 30 min tested following the procedure described by the manufacturer
conditioning sessions following a 15 min pretreatment of morphine (Perkin Elmer, Italia). Displacement curves were generated by
(10 mg/kg, s.c.) (Alajaji et al., 2016). In the PPAR-a antagonism incubating drugs with [3H]-CP-55,940 for 90 min at 30  C. OlGly
study, GW6471 (2 mg/kg, i.p.) was injected 30 min before OlGly. On effect on AEA hydrolysis by rat brain membranes, which express
test day (day 5), mice were allowed access to all chambers for high levels of fatty acid amide hydrolase, were assayed as previ-
15 min in a drug free state. The preference score was calculated by ously described (Ortar et al., 2007).
determining the difference between the time spent in the drug
paired side during test day versus the time in drug paired side 2.11. Molecular modelling studies on OlGly
during the baseline day.
Starting ligand geometry was built with Ghemical 2.99.2
2.8. Tetrad behavioral assessment (Hassinen and Pera €kyla
€, 2001), followed by energy minimization
(EM) at molecular mechanics level first, using Tripos 5.2 force field
Mice were acclimated to the test environment for at least 1 h parametrization, and then at AM1 semi-empirical level. OlGly was
prior to testing for tetrad components: spontaneous activity, cata- fully optimized using GAMESS program at the Hartree-Fock level
lepsy, antinociception, and hypothermia (Little et al., 1988; Martin with STO-3G basis set to derive the partial atomic charges using the
et al., 1991; Wiley and Martin, 2003). Mice were assessed for RESP procedure of restrained fit to the HF/6-31G*/STO-3G electro-
baseline tail withdrawal latencies and body temperature, given an static potential. Docking studies were performed with AutoDock
intraperitoneal (i.p.) injection of vehicle or drug (OlGly), and 30 min 4.2 (Morris et al., 2009). The crystallographic structure of PPAR-a
later assessed in the following order: catalepsy, tail withdrawal test, (PDB entry 2P54) and the ligand were processed with AutoDock
and body temperature. Tools (ADT) package version 1.5.6rc1 (Morris et al., 2009) to merge
In the locomotor studies, subjects were administered vehicle or non polar hydrogens, calculate Gasteiger charges and select rotat-
drug and 5 min later were placed into clear acrylic boxes (approx. able side-chain bonds. Grid for docking evaluation with a spacing of
44.5 cm  22.25 cm  20.0 cm) contained within sound- 0.375 Å and 50  50  70 points, centered in the ligand binding
attenuating cabinets equipped with an LED light source and fans pocket, was generated using the program AutoGrid 4.2 included in
for general air circulation and creation of white noise. The distance Autodock 4.2 distribution. A 100 molecular docking run was per-
traveled was expressed in cm and the time spent immobile in s formed adopting a Lamarckian Genetic Algorithm (LGA) and the
collected and recorded for 10 min using Fire-i™ digital cameras following associated parameters: 100 individuals in a population
purchased from Unibrain (San Ramon, CA, USA) and ANY-maze™ with a maximum of 15 million energy evaluations and a maximum
video tracking software purchased from Stoelting Company (Wood of 37000 generations, followed by 300 iterations of Solis and Wets
Dale, IL, USA). Catalepsy was assessed in the bar test, and the local search. The docking results was subjected to visual inspection
dependent measure was expressed as immobility time during a and as representative binding pose was selected the one with most
60 s observation period (Ignatowska-Jankowska et al., 2015). Anti- favorable binding energy for the subsequent MD simulations of
nociception data were transformed to represent a maximum ligand-PPAR-a complex. The complex was completed by addition of
percent effect (%MPE) by the following formula: %MPE ¼ [(test all hydrogen atoms and underwent EM and then MD simulations
latencypretreatment latency)/(10pretreatment latency)]  100. with Amber12 pmemd.cuda module (Go € tz et al., 2012), using ff12SB
Body temperature data were expressed as a difference between the version of AMBER force field (Case et al., 2012) for the protein and
values collected before ad after the drug or vehicle administration gaff parameters for the ligand.
(D temperature,  C). To perform molecular dynamics (MD) simulation in solvent, the
complex was confined in TIP3P water periodic box exhibiting a
2.9. Cumulative CP55,940 dose-response study minimum distance between solute and box surfaces of 10 Å, using
the tleap module of AmberTools12 program (Wang et al., 2004).
This experiment examined whether OlGly would shift the cu- The system was then neutralized by addition of counterions (Naþ)
mulative dose-response curves of CP55,940 in producing catalepsy, and underwent 1000 steps of EM with solute atoms harmonically
antinociception, and catalepsy, an in vivo assay sensitive to CB1 restrained to their starting positions using a force constant of
receptor allosteric modulators (Ignatowska-Jankowska et al., 2015). 10 kcal mol-1Å-1. The solvated complex was submitted to 90 ps
Mice were pretreated with either OlGly (60 mg/kg i.p.) or vehicle restrained MD (5 kcal mol-1Å-1) at constant volume, gradually
10 min before they received the first dose of CP55,940 followed by heating the system to 300 K, followed by 60 ps restrained MD
each subsequent dose every 40 min. Measurements for catalepsy, in (5 kcal mol-1Å-1) at constant temperature (300 K) and pressure
(bar test), tail-flick, and rectal temperature were taken 30 min (1 atm) to adjust system density. Production MD simulation was
following each CP55,940 administration, as well as prior to any carried out at constant temperature (300 K) and pressure (1 atm)
injections to determine baseline responses. Cumulative doses of for 50 ns with a time-step of 2 fs. Bonds involving hydrogens were
CP55,940 were 0.3, 1, and 3 mg/kg i.p. Locomotor activity was not constrained using the SHAKE algorithm (Ryckaert et al., 1977).
assessed due to habituation effects that occur following repeated
testing. 2.12. PPAR-a luciferase assays

2.10. Cannabinoid receptor binding COS-7 cells (monkey kidney fibroblast-like cells) were grown in
DMEM supplemented with 10% fetal bovine serum and 1% Pen/
Membranes from HEK-293 cells stably transfected with the Strep under standard conditions. Cells were plated in a 24-well
human recombinant CB1 receptor (Bmax ¼ 2.5 pmol/mg protein) plate at confluence and transfected using Lipofectamine LTX and
and human recombinant CB2 receptor (Bmax ¼ 4.7 pmol/mg pro- PLUS Reagent (Life Technologies 15338-100) according to the
tein) were incubated with [3H]-CP-55,940 (0.14 nM/kd ¼ 0.18 nM manufacture's instruction. At day 1, for each well, a combination of
and 0.084 nM/kd ¼ 0.31 nM respectively for CB1 and CB2 receptor) 25 ng of mouse PSG5- PPAR-a (plasmid 22751; Addgene), 300 ng of
324 G. Donvito et al. / Neuropharmacology 148 (2019) 320e331

PPRE X3-TK-luc; (Plasmid 1015 Addgene), 100 ng of pSV-b-Galac- naïve (Fig. 1F) mice. Fig. 2 shows endocannabinoid levels (i.e., AEA,
tosidase Control Vector (Promega E1081) and 75 ng pcDNA3 2-AG) and related N-acyl-ethanolamines levels (i.e., PEA, OEA) in
(Invitrogen) empty vector to a total of 500 ng were transfected. The each of the three brain regions. As shown in Fig. 2A, TBI elicited a
following day, the growth media was replaced with fresh media significant decrease in AEA levels in the insula [F (2,8) ¼ 19.18,
containing compounds listed below and treated overnight. Ethanol p < 0.001] and hippocampus [F (2,8) ¼ 7.587, p < 0.01]. The levels of
was used as vehicle. At day 3 after 18 h of treatment cells were other investigated fatty acid amides in the examined brain regions
harvested and processed for the Luciferase and b -Galactosidase of each experimental group were below detection.
detection analysis.
Luciferase Gene Reporter activity was detected using the Lucif- 3.2. OlGly attenuates nicotine withdrawal and nicotine reward
erase kit (Sigma, LUC1) whereas b-Galactosidase Activity was
detected with the b-Galactosidase detection kit (Sigma, Gal-A). b- Based on the mouse brain injury data revealing a region-
Galactosidase expression was quantified with the Microplate selective increase of OlGly in the insula, we elected to examine
Readers (Tecan) and used as an internal experimental control to whether an i.p. injection of OlGly given 15 min before the nicotine
analyze the transfection efficiencies in each cell sample group. The receptor antagonist mecamylamine would prevent withdrawal
levels of Firefly Luciferase chemiluminescence intensity was signs in nicotine-dependent mice. Whereas nicotine-dependent
detected with a ChemiDoc MP system station using the Imagelab mice undergoing precipitated withdrawal showed a decrease of
software (Biorad) and reported normalized with respect to the b- time spent exploring the open arm in the elevated plus maze assay,
Gal expression. OlGly (30 and 60 mg/kg, i.p.) significantly increased open arm time
[F (3, 56) ¼ 4.661, p < 0.01; Fig. 3A]. Moreover, 60 mg/kg OlGly
2.13. Statistical analysis attenuated the number of mecamylamine-precipitated somatic
withdrawal signs in nicotine-dependent mice [F (3, 56) ¼ 3.439,
Lipid levels are expressed as means ± SEM of pmols/g wet tissue p < 0.01; Fig. 3B], but did not elicit overt behavioral alterations in
weight, unless otherwise stated. One-way ANOVA followed by the control mice continuously infused with saline.
Tukey's test was used for comparisons of AEA, 2-AG, PEA, OEA and Next, we evaluated whether i.p. administration of the most
OlGly levels among the various groups. Unpaired t-test was used to effective dose of OlGly (60 mg/kg) is brain penetrant. Fifteen mi-
analyze the data on the distribution of OlGly in the brain. nutes following the injection, we detected OlGly levels in plasma
In the nicotine withdrawal studies, the data were analyzed by (2004 ± 626.9 pmol/ml), insula (168.0 ± 60.28 pmol/g), hippocam-
two-way ANOVA followed by Holm-Sidak's post-hoc test. For pus (186.0 ± 60.05 pmol/g), and hypothalamus (245.6 ± 108.1 pmol/
conditioned place studies, a preference score was calculated by g). In contrast, OlGly levels were below the limit of quantification in
subtracting time spent in the nicotine-paired chamber post- the vehicle-injected group.
conditioning minus the time spent pre-conditioning. A positive
value indicated a preference for the nicotine- (or morphine-) paired 3.3. OlGly attenuates nicotine-induced CPP
compartment, whereas a negative value indicated an avoidance of
the nicotine- (or morphine-) paired compartment. A number at or We employed the CPP assay, in order to test whether OlGly re-
near zero indicated no preference. Data were analyzed by one-way duces the rewarding effects of nicotine. OlGly dose-dependently
ANOVA and further analyzed by the Student Neuman-Keuls post- prevented the development of nicotine-induced CPP [F
hoc test. (7,55) ¼ 12.77, p < 0.001; Fig. 4A], but did not affect place prefer-
In tetrad studies and luciferase assay, data were analyzed by ence in saline-treated mice. However, the OlGly reduction of
one-way ANOVA followed by the Dunnett's post-hoc test. In the nicotine CPP did not carry over to morphine CPP (Fig. 4B), sug-
cumulative dose-response of CP55,940, data were analyzed by two- gesting that OlGly is comparatively selective in blocking nicotine
way ANOVA followed by the Sidak post-hoc test. In the luciferase CPP.
assay, the Student's t-test with Welch's correction was applied. In
the binding studies, Ki values were calculated by applying the 3.4. OlGly does not functionally interact with the endogenous
Cheng-Prusoff equation to the IC50 values for the displacement of cannabinoid system
the bound radioligand by increasing concentrations of the test
compound. Data are expressed as means ± SEM of at least n ¼ 3 Because the endogenous cannabinoid system modulates
experiments. numerous indices of nicotine reward and dependence, the next
The computer program GraphPad Prism version 6.0 (GraphPad series of experiments investigated OlGly in a variety of in vivo and
Software Inc., San Diego, CA) was used in all statistical analyses. All in vitro assays indicative of cannabimimetic activity. OlGly (10, 30,
data are expressed as mean ± SEM. A P value of <0.05 was and 100 mg/kg i.p.) did not elicit significant effects in the tetrad
considered statistically significant. assay, a battery of in vivo measures highly associated with CB1 re-
ceptor activity (Martin et al., 1991). Specifically, OlGly did not elicit
3. Results catalepsy in the bar test, antinociceptive effects in the tail-flick test,
hypothermia, or hypomotility (Table 1). Additionally, OlGly did not
3.1. OlGly levels are increased in the insular cortex of TBI mice alter the dose-response relationship of the high efficacy CB1 re-
ceptor agonist CP55,940 (Devane et al., 1988) in the tetrad assay
Experimental TBI increased OlGly levels, but not other analyzed (Fig. 5), suggesting that it does not act functionally as a CB1 receptor
lipids, in the insular cortex of mice [F (2,10) ¼ 15.46, p < 0.001; antagonist or CB1 receptor allosteric modulator. Finally, OlGly did
Fig. 1A] compared to sham and naïve mice. However, OlGly levels not bind CB1 or CB2 receptors and weakly inhibited FAAH (Table 2).
did not significantly differ between brain-injured and sham groups
in the hippocampus (Fig. 1B) or hypothalamus (Fig. 1C), indicating 3.5. OlGly elicits its effects via PPAR-a mechanism of action
regional selectivity. The chromatograms show the presence of
OlGly in the insula of TBI mice (Fig. 1D), but not in sham (Fig. 1E) or Considering that OlGly possesses a chemical structure similar to
G. Donvito et al. / Neuropharmacology 148 (2019) 320e331 325

Fig. 1. Traumatic brain injury (TBI) leads to increased OlGly levels in the insular cortex of mice. (A) Mice subjected to TBI display significant increases of OlGly in the insula, but not in
(B) hippocampus or (C) hypothalamus. Values represent the mean ± SEM of n ¼ 5. **p < 0.001 vs naïve; ##p < 0.001 vs sham. (D) A representative chromatogram shows the
presence of OlGly in the insula of mice 24 h following TBI. Injured insula shows formation of OlGly as confirmed by MS and MS/MS spectra. However, OlGly was below the limit of
detection in sham (E) and naïve mice (F). The arrow depicts the retention time of OlGly from the standard. The upper chromatogram traces represent the total ion current (TIC), and
lower chromatogram traces represent the extracted chromatograms m/z around 340 amu in (D-F).

OEA, which acts as an endogenous PPAR-a ligand, we next assessed selective PPAR-a antagonist GW6471 fully prevented OlGly
whether OlGly binds PPAR-a as well as produces functional activity blockade of nicotine CPP [F (6, 56) ¼ 12.8, p < 0.0001].
at this receptor. As shown in the modeling experiment, OlGly binds
PPAR-a (Fig. 6A), and behaved as a PPAR-a receptor agonist in a
specific luciferase assay for functional activity (Fig. 6B). The selec- 4. Discussion
tive PPAR-a agonist GW7647 (comparison drug), as well as OlGly,
significantly increased luciferase activity compared to DMSO [F The impetus of the present study was based on a report that
(5,44) ¼ 17.7, p < 0.0001]. nicotine addiction was ameliorated in cigarette smokers suffering
Given this in vitro evidence suggesting that OlGly behaves as a from brain damage that included the insular cortex compared with
PPAR-a receptor agonist, we next examined whether PPAR-a me- brain-damaged individuals that did not involve the insula, sug-
diates OlGly prevention of nicotine CPP. As shown in Fig. 6C, the gesting that this brain region may play an important role in
smoking addiction (Naqvi et al., 2014, 2007). We speculated that
326 G. Donvito et al. / Neuropharmacology 148 (2019) 320e331

Fig. 2. Levels of endocannabinoids in the hippocampus, hypothalamus and insula of sham mice and mice following weight drop-induced traumatic brain injury. (A) Levels of
anandamide (AEA) and 2-arachidonoylglycerol (2-AG). (B) Palmitoylethanolamide (PEA) and oleoylethanolamide (OEA) levels. Values represent means ± SEM of n ¼ 4e5 ex-
periments. **p < 0.001, *p < 0.05 vs naïve; ##p < 0.001, #p < 0.05 vs sham.

Fig. 3. OlGly prevents mecamylamine-precipitated withdrawal signs in nicotine-dependent mice. Intraperitoneal administration of OlGly significantly (A) increased time spent in
the open arms of the elevated plus maze compared to nicotine-pelleted mice that were given an injection of vehicle, and (B) decreased total somatic signs. All mice were challenged
with mecamylamine (2 mg/kg, s.c.) prior to testing. MP: minipump. Values represent the mean ± SEM of n ¼ 7e8 mice per group. *p < 0.05 vs. saline/vehicle; #p < 0.05 vs. nicotine/
vehicle.

experimental brain injury in mice might alter neurochemicals in whether its exogenous administration would ameliorate
this brain region that may serve as a starting point for identifying withdrawal-associated behaviors in nicotine-dependent mice as
new molecules to treat nicotine addiction. Accordingly, we exam- well prevent nicotine reward in the mouse CPP model. Three gen-
ined the lipidomic profile of the insular cortex in mice subjected to eral findings support the idea that OlGly may serve to counteract
TBI and found a profound increase of a largely uncharacterized N- nicotine addiction. First, OlGly reduced somatic withdrawal signs as
acylglycine, OlGly, in the insular cortex, but not in hippocampus or well as affective behavior in nicotine-dependent mice undergoing
hypothalamus. N-acylglycines represent a possible class of endog- precipitated withdrawal. Second, OlGly prevented the development
enous signaling molecules, which are structurally related to of nicotine CPP. Third, a singular systemic administration of the
endocannabinoids, and have recently generated research interest most effective dose of OlGly in naïve mice results in an increase of
due to their remarkable biological activities, such as anti- OlGly contents within plasma, and in the brain to levels similar to
inflammatory effects (Burstein et al., 2011), inhibition of cancer those found in mice after TBI.
cell proliferation (Chatzakos et al., 2012), adipogenesis (Wang et al., Few studies have investigated the pharmacological effects of
2015), and neural protection (Cohen-Yeshurun et al., 2011). Based OlGly. Although it has been detected in different tissues, including
on this regionally selective increase of OlGly, we sought to test rat spinal cord, brain, lung, skin, ovaries, liver, spleen, and kidney
G. Donvito et al. / Neuropharmacology 148 (2019) 320e331 327

Fig. 4. OlGly prevents nicotine reward, but not morphine reward. (A) OlGly dose-dependently blocked nicotine CPP, (B) but did not attenuate morphine (30 mg/kg, i.p.) CPP. Values
represent the mean ± SEM of n ¼ 7e8 mice per group. *p < 0.05 vs. saline/vehicle; #p < 0.05 vs. nicotine/vehicle.

Table 1
OlGly does not elicit in vivo pharmacological effects as assessed in the tetrad assay. OlGly did not produce catalepsy, antinociception, hypothermia, or motor behavior, as
reflected by the following measures: distance traveled, speed, and immobility time. Values represent means ± SEM of n ¼ 9 mice per group.

Treatment Catalepsy (s) Antinociception (%MPE) D rectal temperature ( C) Distance traveled (cm) Speed (cm/s) Time immobile (s)

vehicle 0.0 6.0 ± 5.1 0.41 ± 0.30 20.8 ± 2.3 6.9 ± 0.8 7.1 ± 1.5
OlGly 10 mg/kg 0.0 12.0 ± 6.6 0.90 ± 0.19 24.3 ± 1.2 8.1 ± 0.4 8.6 ± 1.6
OlGly 30 mg/kg 0.0 6.0 ± 4.5 1.13 ± 0.26 21.5 ± 1.7 7.2 ± 0.6 11.8 ± 6.2
OlGly 100 mg/kg 0.0 25.9 ± 11.0 0.22 ± 0.5431 18.6 ± 1.5 6.21 ± 0.5 17.4 ± 7.3

Fig. 5. OlGly does not act as a CB1 receptor allosteric modulator in the in vivo triad assay. CP55,940 (0, 0.3, 1, and 3 mg/kg) dose-dependently induced (A) catalepsy, (B) anti-
nociception, and (C) hypothermia in vehicle-pretreated mice. Pretreatment with OlGly (60 mg/kg) did not induce any changes in the dose-response curve of CP55,940 for any of
these dependent measures. Values represent means ± SEM of n ¼ 5/6 mice per group.

Table 2
OlGly does not functionally interact with the endogenous cannabinoid system. OlGly shows low affinity for CB1 and CB2 receptors and has very low potency in inhibiting FAAH.
Values represent mean ± SEM (n ¼ 3), % displacement or inhibition, n ¼ 2.

CB1 IC50 CB1 Ki CB1 displacement produced by CB2 IC50 CB2 Ki CB2 displacement produced by IC50 FAAH FAAH inhibition produced by
(mM) (mM) maximum dose tested (50 mM) (mM) (mM) maximum dose tested (50 mM) inhibition (mM) maximum dose tested (50 mM)

45.5 ± 5.1 27.8 ± 3.1 55.9 ± 7.3% >50 >50 15.5 ± 6.40% 8.65 89.4%

(Bradshaw et al., 2009), the present study is the first to quantify it in pharmacological effects occur independently of its conversion to
specific brain regions. OlGly biosynthesis results from a reaction oleamide. In addition to its structural similarity to the N-acyle-
between oleoyl CoA and glycine, which can be catalysed by either thanolamines, which include AEA, as well as PPAR-a agonists OEA
glycine N-acyltransferase-like 3, or cytochrome c in the presence of and palmitoylethanolamide (PEA), recent evidence suggests that
hydrogen peroxide (Wang et al., 2015), while FAAH participates in OlGly may play a role in enhancing insulin sensitivity through a CB1
its degradation (Bradshaw et al., 2009). Additionally, OlGly may receptor mechanism of action (Wang et al., 2015). The CB1 receptor
play a role in the biosynthesis of oleamide, another substrate of also plays an important role in mediating the rewarding effects of
FAAH that mediates several fundamental neurochemical processes nicotine. For example, a CB1 receptor antagonist administered
(Boger et al., 1998) including sleep (Huitron-Resendiz et al., 2004), either systemically or into the ventral tegmental area (VTA)
thermoregulation, and nociception (Fedorova et al., 2001). OlGly decreased nicotine self-administration in rats (Le Foll and Goldberg,
and oleamide possess equal potency in eliciting hypothermia and 2004; Simonnet et al., 2013). Similarly, CB1 (/) mice or wild type
decreasing locomotion in rats. However, the failure of OlGly to in- animals treated with a selective CB1 receptor antagonist do not
crease circulating levels of oleamide suggests that its display nicotine CPP (Castan~e et al., 2002; Le Foll and Goldberg,
328 G. Donvito et al. / Neuropharmacology 148 (2019) 320e331

Fig. 6. PPAR-a mediates the anti-reward effects of OlGly. (A) Representative frame from MD of OlGly/PPAR-a complex and details of ligand-protein interactions in the ligand binding
site. The carboxylic moiety of the ligand recapitulates the main polar stabilizing interactions with Tyr464(H12), Tyr314(H5), His440(H10/11) and Ser280(H3), signature of a PPAR-a
agonist. (B) Luciferase Assay for PPAR-a/RXR. Relative Luciferase Units in response to OlGly and the PPAR-a agonist GW7647 (comparison drug). DMSO (n ¼ 12), GW7647 (GW)
10 mM (n ¼ 6), OlGly 10 mM (n ¼ 7), 50 mM (n ¼ 9) and 100 mM (n ¼ 11). ****p < 0.0001, *p < 0.05 vs DMSO (C) The selective PPAR-a antagonist GW6471 (2 mg/kg) prevented OlGly-
induced inhibition of nicotine CPP. n ¼ 6e8 mice per group. ****p < 0.0001. ***p < 0.001, **p < 0.01 vs. vehicle/vehicle; ###p < 0.001 vs. nicotine/vehicle. Values represent the
mean ± SEM.

2004; Merritt et al., 2008), indicating that this receptor plays a inhibitors reduce nicotine self-administration and nicotine
necessary role in multiple laboratory models of nicotine reward. priming-induced reinstatement through PPAR-a in squirrel mon-
Likewise, genetic deletion or pharmacological inhibition of CB2 keys, consistent with the idea that the endogenous substrates of
receptors blocks nicotine reward-like effects in the nicotine self- this enzyme counteract nicotine reward by activating this nuclear
administration and nicotine CPP paradigms in mice (Ignatowska- receptor (Justinova et al., 2015). In contrast, Forget and colleagues
Jankowska et al., 2013; Navarrete et al., 2013). In contrast, a CB2 have found that inhibition of FAAH reduced nicotine reinstatement,
receptor antagonist did not affect either nicotine self- but did not affect nicotine self-administration (Forget et al., 2009).
administration or reinstatement of nicotine seeking in rats Accordingly, our experiment showing that OlGly only weakly in-
(Gamaleddin et al., 2012). These divergent results of the effective- hibits AEA hydrolysis suggests the unlikelihood of indirect activa-
ness of CB2 receptor antagonists in reducing nicotine reward may tion of cannabinoid receptors or other AEA targets through FAAH
be related to species differences. Importantly, clinical trials inhibition.
revealed the CB1 receptor antagonist rimonabant effectively Structural similarities between OlGly and OEA, which activates
improved smoking abstinence compared with placebo, though PPAR-a, raise the possibility that OlGly may likewise prevent the
clinical development of this and other CB1 receptor antagonists was development of nicotine CPP through this receptor. Notably, PPAR-a
terminated due to unacceptable adverse effects (Robinson et al., has been implicated in the control of nicotine reward (Jackson et al.,
2017). The question of whether a CB2 receptor antagonist has effi- 2017; Mascia et al., 2011; Panlilio et al., 2012). The modeling
cacy in treating smoking addiction remains to be determined. experiment presented here revealing that OlGly binds PPAR-a and
Based on the extensive role that the endogenous cannabinoid the finding that OlGly behaved as a PPAR-a agonist in a specific
system plays on nicotine reward, we tested whether OlGly interacts luciferase assay support this hypothesis. Another noteworthy
with this system. OlGly did not produce in vivo pharmacological finding in the present study is that the selective PPAR-a antagonist
activity indicative of CB1 receptor stimulation as assessed in the GW6471 fully prevented OlGly blockade of nicotine CPP, indicating
tetrad assay, consisting of catalepsy, antinociception, hypothermia, that this anti-reward effect of OlGly requires PPAR-a receptor
and hypomotility measures (Little et al., 1988; Martin et al., 1991; activation. These results are consistent with recent reports on
Wiley and Martin, 2003). In contrast, Chaturvedi and colleagues PPAR-a agonists blocking nicotine withdrawal, nicotine CPP, nico-
reported that OlGly reduces body temperature (1.5e2.0  C) and tine self-administration, and nicotine priming-induced reinstate-
decreases locomotor activity in rats (Chaturvedi et al., 2006). The ment in rodents and nonhuman primates (Jackson et al., 2017;
disparate results between their finds and the results in the present Mascia et al., 2011; Panlilio et al., 2012). However, fenofibrate, a low
study may be a consequence of species differences or other efficacy and non-selective PPAR-a agonist, did not facilitate the
methodological considerations (e.g., housing of the animals, ability to stop smoking during a brief practice quit period in
ambient room temperature, etc.). At any rate, the previous study dependent smokers (Perkins et al., 2016). In addition, it failed to
did not examine CB1 receptor involvement of the hypothermic or reverse nicotine withdrawal in mice (Jackson et al., 2017). None-
decreases locomotor activity effects of OlGly. Furthermore, the fact theless, clinical studies employing PPAR-a agonists with higher
that OlGly poorly binds human recombinant CB1 and CB2 receptors efficacy and selectivity remain to be conducted.
suggests direct action at these receptors is unlikely. Another pos- The findings that OlGly dose-dependently reduced the devel-
sibility is that OlGly behaves as an allosteric modulator of the CB1 opment of nicotine-induced CPP, but did not affect morphine CPP,
receptor. Accordingly, we examined whether OlGly would alter the suggest selectivity for blocking nicotine CPP. This observation is
antinociceptive, cataleptic, and hypothermic effects of the high consistent with work reviewed by Melis and Pistis (2014), who
efficacy, orthosteric CB1 receptor agonist CP55,940. Whereas the reported that the “anti-addictive” properties of PPAR-a stimulation
CB1 receptor positive allosteric modulator ZCZ011 elicited a left- show selectivity for reducing nicotine reward over the reinforcing
ward shift of the CP55,940 dose-response curve for each of these effects of cocaine, cannabinoids, and morphine (Melis and Pistis,
measures (Ignatowska-Jankowska et al., 2015), OlGly did not affect 2014). A parsimonious explanation accounting for the selectivity
the CP55,940 dose-response curves. It is noteworthy that FAAH of OlGly in blocking nicotine CPP includes a complex
G. Donvito et al. / Neuropharmacology 148 (2019) 320e331 329

neurochemical pathway involving PPAR-a activity inducing nega- attenuate nicotine reinforcement and reinstatement in rodents and
tive modulation of b2*-nAChR receptor activity expressed on VTA nonhuman primates (Mascia et al., 2011; Panlilio et al., 2012),
dopamine cells that ultimately dampen nicotine-induced firing and though the neuroanatomical substrates remain to be determined as
bursting activity (Melis et al., 2013). Similarly, fenofibrate induced a well as the potential translational efficacy in humans. Nonetheless,
reduction of stress-induced depression behaviors in mice through the findings that i.p. administration of OlGly prevented both nico-
phasic activation of the mesolimbic dopaminergic system (Scheggi tine CPP and nicotine withdrawal-associated behaviors in mice, and
et al., 2016). Additionally, it has been reported that local infusion of that the injection of the same effective dose of OlGly in naïve mice
dopamine receptor 1 antagonist into the insular cortex significantly was brain penetrant suggest that this ligand may be of value in
decreased nicotine self-administration in rats (Kutlu et al., 2013). treating addiction to tobacco smoking in humans.
According to the model proposed by Naqvi and Bechera, the
insula belongs to a network, which includes the ventromedial Funding
prefrontal cortex, anterior cingulate cortex, NAc, and amygdala,
responsible for the conscious pleasure from drugs, drug craving, This work was supported by NIH grants P01DA009789,
drug seeking behavior, and drug relapse. Specifically, the insula R01DA039942, P30DA033934 (AHL); R01 DA032246 and
processes interoceptive information about the drugs received from P50DA039841 (MID), T32DA007027 (AJ), and startup funds from
a thalamocortical pathway and relays this information to the pre- the VCU School of Pharmacy. NSERC (92056) and CIHR (137122)
frontal regions and the amygdala, which evoke pleasure related grants to LAP.
effects of the drugs through dopaminergic activation in the VTA
(Naqvi and Bechara, 2010). According to their model, the insula Conflicts of interest
serves as a critical neural substrate for addiction by regulating the
dopaminergic signal in the VTA. However, whether a direct None.
connection exists between the mesolimbic dopamine system and
PPAR-aeinduced reduction of nicotine CPP by OlGly remains an Authorship contributions
unanswered question. It will also be important to determine
whether the anti-nicotine CPP effects of OlGly extend to self- Participated in the research design: Donvito, Piscitelli, Petrie,
administration and reinstatement procedures. Parker, Maione, Lichtman, Damaj, Di Marzo, Mechoulam.
We based the rationale to investigate the effects of OlGly on Conducted experiments: Piscitelli, Muldoon, Jackson, Vitale,
nicotine CPP and withdrawal responses in nicotine-dependent D'Aniello, Giordano, Guida, Ignatowska-Jankowska, Mustafa, Sim-
mice from lipidomic analyses of the insular cortex from mice Selley.
following TBI. Although the present study does not address the Contributed to reagents or analytical tool: Smoum.
neural substrates underlying these effects, we found that following Performed data analysis: Donvito, Piscitelli, Muldoon, Jackson,
a singular i.p. administration of a dose of OlGly that was most D'Aniello, Mustafa, Lichtman, Damaj, Di Marzo.
effective in reducing nicotine withdrawal, the levels of OlGly Wrote or contributed to the writing of the manuscript: Donvito,
increased within the insula, hippocampus, hypothalamus and Piscitelli, Lichtman, Damaj, Di Marzo, Mechoulam.
plasma up to concentrations that were similar to those detected
after TBI. Nonetheless, other preclinical research supports a role of Declarations of interest
the insular cortex on nicotine reward. Whereas inactivation of
insula by local infusion of geaminobutyric acid agonist mixture Drs. Damaj, Di Marzo, Lichtman, Mechoulam, and Parker filed a
decreased nicotine self-administration in rats (Forget et al., 2010; provisional patent on oleoyl glycine.
Pushparaj et al., 2015), electrical stimulation of this region signifi-
cantly attenuated nicotine reinforcement as well as nicotine- References
seeking behaviors. These findings suggest that the insular cortex
may play a modulatory role in nicotine reward (Pushparaj et al., Abdolahi, A., Acosta, G., Breslin, F.J., Hemby, S.E., Lynch, W.J., 2010. Incubation of
nicotine seeking is associated with enhanced protein kinase A-regulated
2013). In the case of dependence, both central and peripheral
signaling of dopamine- and cAMP-regulated phosphoprotein of 32 kDa in the
nicotinic receptors mediate somatic nicotine withdrawal signs, insular cortex. Eur. J. Neurosci. 31, 733e741. https://doi.org/10.1111/j.1460-
while central nicotinic receptor populations, such as the nucleus 9568.2010.07114.x.
accumbens (NAc) and habenulo-interpeduncular system regulate Alajaji, M., Lazenka, M.F., Kota, D., Wise, L.E., Younis, R.M., Carroll, F.I., Levine, A.,
Selley, D.E., Sim-Selley, L.J., Damaj, M.I., 2016. Early adolescent nicotine expo-
anhedonia, anxiety, and aversion (De Biasi and Dani, 2011; Watkins sure affects later-life cocaine reward in mice. Neuropharmacology 105,
et al., 2000). Furthermore, the mesohabenular pathway through 308e317. https://doi.org/10.1016/j.neuropharm.2016.01.032.
the interpeduncular nucleus and corticotropin-releasing factor-1 Albuquerque, E.X., Pereira, E.F.R., Alkondon, M., Rogers, S.W., 2009. Mammalian
nicotinic acetylcholine receptors: from structure to function. Physiol. Rev. 89,
(CRF1) receptors in the central nucleus of the amygdala (CeA) seem 73e120. https://doi.org/10.1152/physrev.00015.2008.
to play a preponderant role in the emergence of anxiety-like be- Bisogno, T., Piscitelli, F., Di Marzo, V., 2009. Lipidomic methodologies applicable to
haviors in nicotine-dependent animals (Cohen et al., 2015). Thus, the study of endocannabinoids and related compounds: Endocannabinoido-
mics. Eur. J. Lipid Sci. Technol. 111, 53e63. https://doi.org/10.1002/
the neural sites of action of OlGly and whether the elevated levels of ejlt.200800233.
OlGly in the insular cortex produced by TBI ameliorates nicotine Boger, D.L., Patterson, J.E., Guan, X., Cravatt, B.F., Lerner, R.A., Gilula, N.B., 1998.
reward and/or dependence remain to be determined. Chemical requirements for inhibition of gap junction communication by the
biologically active lipid oleamide. Proc. Natl. Acad. Sci. U. S. A 95, 4810e4815.
Bradshaw, H.B., Rimmerman, N., Hu, S.S.-J., Burstein, S., Walker, J.M., 2009. Novel
5. Conclusions endogenous N-acyl glycines identification and characterization. Vitam. Horm.
81, 191e205. https://doi.org/10.1016/S0083-6729(09)81008-X.
Burstein, S.H., McQuain, C.A., Ross, A.H., Salmonsen, R.A., Zurier, R.E., 2011. Reso-
The present study demonstrates an alteration in the lipid profile
lution of inflammation by N-arachidonoylglycine. J. Cell. Biochem. 112,
in the insular cortex compared to other neural regions of mice 3227e3233. https://doi.org/10.1002/jcb.23245.
subjected to TBI. In particular, we found a significant increase of Case, D., Darden, T., Cheatham III, T., Simmerling, C., Wang, J., Duke, R., Luo, R.,
OlGly levels in the insula, a region implicated in smoking addiction Walker, R., Zhang, W., Merz, K., Roberts, B., Hayik, S., Roitberg, A., Seabra, G.,
€ tz, A., Kolossva
Swails, J., Go ry, I., Wong, K., Paesani, F., Vanicek, J., Wolf, R., Liu, J.,
(Naqvi et al., 2014, 2007). A potential PPAR-a mechanism of action Wu, X., Brozell, S., Steinbrecher, T., Gohlke, H., Cai, Q., Ye, X., Wang, J., Hsieh, M.,
is consistent with several reports showing that PPAR-a agonists Cui, G., Roe, D., Mathews, D., Seetin, M., Salomon-Ferrer, R., Sagui, C., Babin, V.,
330 G. Donvito et al. / Neuropharmacology 148 (2019) 320e331

Luchko, T., Gusarov, S., Kovalenko, A., Kollman, P., 2012. Amber 12. Jackson, K.J., Martin, B.R., Changeux, J.P., Damaj, M.I., 2008. Differential role of
Castan~e
, A., Berrendero, F., Maldonado, R., 2005. The role of the cannabinoid system nicotinic acetylcholine receptor subunits in physical and affective nicotine
in nicotine addiction. Pharmacol. Biochem. Behav. 81, 381e386. https://doi.org/ withdrawal signs. J. Pharmacol. Exp. Therapeut. 325, 302e312. https://doi.org/
10.1016/j.pbb.2005.01.025. 10.1124/jpet.107.132977.
Castan~e
, A., Valjent, E., Ledent, C., Parmentier, M., Maldonado, R., Valverde, O., 2002. Justinova, Z., Panlilio, L.V., Moreno-Sanz, G., Redhi, G.H., Auber, A., Secci, M.E.,
Lack of CB1 cannabinoid receptors modifies nicotine behavioural responses, but Mascia, P., Bandiera, T., Armirotti, A., Bertorelli, R., Chefer, S.I., Barnes, C.,
not nicotine abstinence. Neuropharmacology 43, 857e867. Yasar, S., Piomelli, D., Goldberg, S.R., 2015. Effects of fatty acid amide hydrolase
Chaturvedi, S., Driscoll, W.J., Elliot, B.M., Faraday, M.M., Grunberg, N.E., Mueller, G.P., (FAAH) inhibitors in non-human primate models of nicotine reward and
2006. In vivo evidence that N-oleoylglycine acts independently of its conver- relapse. Neuropsychopharmacology 40, 2185e2197. https://doi.org/10.1038/
sion to oleamide. Prostag. Other Lipid Mediat. 81, 136e149. https://doi.org/ npp.2015.62.
10.1016/j.prostaglandins.2006.09.001. Kota, D., Martin, B.R., Robinson, S.E., Damaj, M.I., 2007. Nicotine dependence and
Chatzakos, V., Sla €tis, K., Djureinovic, T., Helleday, T., Hunt, M.C., 2012. N-acyl tau- reward differ between adolescent and adult male mice. J. Pharmacol. Exp.
rines are anti-proliferative in prostate cancer cells. Lipids 47, 355e361. https:// Therapeut. 322, 399e407. https://doi.org/10.1124/jpet.107.121616.
doi.org/10.1007/s11745-011-3639-9. Kutlu, M.G., Burke, D., Slade, S., Hall, B.J., Rose, J.E., Levin, E.D., 2013. Role of insular
Cohen-Yeshurun, A., Trembovler, V., Alexandrovich, A., Ryberg, E., Greasley, P.J., cortex D1 and D2 dopamine receptors in nicotine self-administration in rats.
Mechoulam, R., Shohami, E., Leker, R.R., 2011. N-arachidonoyl-L-serine is neu- Behav. Brain Res. 256, 273e278. https://doi.org/10.1016/j.bbr.2013.08.005.
roprotective after traumatic brain injury by reducing apoptosis. J. Cerebr. Blood Le Foll, B., Goldberg, S.R., 2004. Rimonabant, a CB1 antagonist, blocks nicotine-
Flow Metabol. 31, 1768e1777. https://doi.org/10.1038/jcbfm.2011.53. conditioned place preferences. Neuroreport 15, 2139e2143.
Cohen, A., Treweek, J., Edwards, S., Lea ~o, R.M., Schulteis, G., Koob, G.F., George, O., Little, P.J., Compton, D.R., Johnson, M.R., Melvin, L.S., Martin, B.R., 1988. Pharma-
2015. Extended access to nicotine leads to a CRF1 receptor dependent increase cology and stereoselectivity of structurally novel cannabinoids in mice.
in anxiety-like behavior and hyperalgesia in rats. Addiction Biol. 20, 56e68. J. Pharmacol. Exp. Therapeut. 247, 1046e1051.
https://doi.org/10.1111/adb.12077. Martin, B.R., Compton, D.R., Thomas, B.F., Prescott, W.R., Little, P.J., Razdan, R.K.,
De Biasi, M., Dani, J.A., 2011. Reward, addiction, withdrawal to nicotine. Annu. Rev. Johnson, M.R., Melvin, L.S., Mechoulam, R., Ward, S.J., 1991. Behavioral,
Neurosci. 34, 105e130. https://doi.org/10.1146/annurev-neuro-061010-113734. biochemical, and molecular modeling evaluations of cannabinoid analogs.
Devane, W.A., Dysarz, F.A., Johnson, M.R., Melvin, L.S., Howlett, A.C., 1988. Deter- Pharmacol. Biochem. Behav. 40, 471e478.
mination and characterization of a cannabinoid receptor in rat brain. Mol. Mascia, P., Pistis, M., Justinova, Z., Panlilio, L.V., Luchicchi, A., Lecca, S., Scherma, M.,
Pharmacol. 34, 605e613. Fratta, W., Fadda, P., Barnes, C., Redhi, G.H., Yasar, S., Le Foll, B., Tanda, G.,
Devane, W.A., Hanus, L., Breuer, A., Pertwee, R.G., Stevenson, L.A., Griffin, G., Piomelli, D., Goldberg, S.R., 2011. Blockade of nicotine reward and reinstatement
Gibson, D., Mandelbaum, A., Etinger, A., Mechoulam, R., 1992. Isolation and by activation of alpha-type peroxisome proliferator-activated receptors. Biol.
structure of a brain constituent that binds to the cannabinoid receptor. Science Psychiatr. 69, 633e641. https://doi.org/10.1016/j.biopsych.2010.07.009.
258, 1946e1949. Mechoulam, R., Ben-Shabat, S., Hanus, L., Ligumsky, M., Kaminski, N.E., Schatz, A.R.,
Doll, R., Peto, R., Boreham, J., Sutherland, I., 2004. Mortality in relation to smoking: Gopher, A., Almog, S., Martin, B.R., Compton, D.R., Pertwee, R.G., Griffin, G.,
50 years' observations on male British doctors. BMJ 328, 1519. https://doi.org/ Bayewitch, M., Barg, J., Vogel, Z., 1995. Identification of an endogenous 2-
10.1136/bmj.38142.554479.AE. monoglyceride, present in canine gut, that binds to cannabinoid receptors.
Fedorova, I., Hashimoto, A., Fecik, R.A., Hedrick, M.P., Hanus, L.O., Boger, D.L., Biochem. Pharmacol. 50, 83e90. https://doi.org/10.1016/0006-2952(95)00109-
Rice, K.C., Basile, A.S., 2001. Behavioral evidence for the interaction of oleamide D.
with multiple neurotransmitter systems. J. Pharmacol. Exp. Therapeut. 299, Melis, M., Pistis, M., 2014. Targeting the interaction between fatty acid ethanola-
332e342. mides and nicotinic receptors: therapeutic perspectives. Pharmacol. Res. 86,
Forget, B., Coen, K.M., Le Foll, B., 2009. Inhibition of fatty acid amide hydrolase 42e49. https://doi.org/10.1016/j.phrs.2014.03.009.
reduces reinstatement of nicotine seeking but not break point for nicotine self- Melis, M., Scheggi, S., Carta, G., Madeddu, C., Lecca, S., Luchicchi, A., Cadeddu, F.,
administrationdcomparison with CB1 receptor blockade. Psychopharmacology Frau, R., Fattore, L., Fadda, P., Ennas, M.G., Castelli, M.P., Fratta, W., Schilstrom, B.,
(Berl) 205, 613e624. https://doi.org/10.1007/s00213-009-1569-5. Banni, S., De Montis, M.G., Pistis, M., 2013. PPARa regulates cholinergic-driven
Forget, B., Pushparaj, A., Le Foll, B., 2010. Granular insular cortex inactivation as a activity of midbrain dopamine neurons via a novel mechanism involving a7
novel therapeutic strategy for nicotine addiction. Biol. Psychiatr. 68, 265e271. nicotinic acetylcholine receptors. J. Neurosci. 33, 6203e6211. https://doi.org/
https://doi.org/10.1016/j.biopsych.2010.01.029. 10.1523/JNEUROSCI.4647-12.2013.
Gamaleddin, I., Zvonok, A., Makriyannis, A., Goldberg, S.R., Le Foll, B., 2012. Effects of Merritt, L.L., Martin, B.R., Walters, C., Lichtman, A.H., Damaj, M.I., 2008. The
a selective cannabinoid CB2 agonist and antagonist on intravenous nicotine self endogenous cannabinoid system modulates nicotine reward and dependence.
administration and reinstatement of nicotine seeking. PLoS One 7. https:// J. Pharmacol. Exp. Therapeut. 326, 483e492. https://doi.org/10.1124/
doi.org/10.1371/journal.pone.0029900 e29900. jpet.108.138321.
Gamaleddin, I.H., Trigo, J.M., Gueye, A.B., Zvonok, A., Makriyannis, A., Goldberg, S.R., Morris, G.M., Huey, R., Lindstrom, W., Sanner, M.F., Belew, R.K., Goodsell, D.S.,
Le Foll, B., 2015. Role of the endogenous cannabinoid system in nicotine Olson, A.J., 2009. AutoDock4 and AutoDockTools4: automated docking with
addiction: novel insights. Front. Psychiatr. 6, 41. https://doi.org/10.3389/ selective receptor flexibility. J. Comput. Chem. 30, 2785e2791. https://doi.org/
fpsyt.2015.00041. 10.1002/jcc.21256.
Go€tz, A.W., Williamson, M.J., Xu, D., Poole, D., Le Grand, S., Walker, R.C., 2012. Naqvi, N.H., Bechara, A., 2010. The insula and drug addiction: an interoceptive view
Routine microsecond molecular dynamics simulations with AMBER on GPUs. 1. of pleasure, urges, and decision-making. Brain Struct. Funct. 214, 435e450.
Generalized born. J. Chem. Theor. Comput. 8, 1542e1555. https://doi.org/ https://doi.org/10.1007/s00429-010-0268-7.
10.1021/ct200909j. Naqvi, N.H., Gaznick, N., Tranel, D., Bechara, A., 2014. The insula: a critical neural
Hassinen, T., Pera €kyla €, M., 2001. New energy terms for reduced protein models substrate for craving and drug seeking under conflict and risk. Ann. N. Y. Acad.
implemented in an off-lattice force field. J. Comput. Chem. 22, 1229e1242. Sci. 1316, 53e70. https://doi.org/10.1111/nyas.12415.
https://doi.org/10.1002/jcc.1080. Naqvi, N.H., Rudrauf, D., Damasio, H., Bechara, A., 2007. Damage to the insula dis-
Heishman, S.J., Kleykamp, B.A., Singleton, E.G., 2010. Meta-analysis of the acute rupts addiction to cigarette smoking. Science 315, 531e534. https://doi.org/
effects of nicotine and smoking on human performance. Psychopharmacology 10.1126/science.1135926.
(Berl) 210, 453e469. https://doi.org/10.1007/s00213-010-1848-1. Navarrete, F., Rodríguez-Arias, M., Martín-García, E., Navarro, D., García-
Huitron-Resendiz, S., Sanchez-Alavez, M., Wills, D.N., Cravatt, B.F., Henriksen, S.J., Gutierrez, M.S., Aguilar, M.A., Aracil-Fern andez, A., Berbel, P., Min ~ arro, J.,
2004. Characterization of the sleep-wake patterns in mice lacking fatty acid Maldonado, R., Manzanares, J., 2013. Role of CB2 cannabinoid receptors in the
amide hydrolase. Sleep 27, 857e865. rewarding, reinforcing, and physical effects of nicotine. Neuro-
Ignatowska-Jankowska, B.M., Baillie, G.L., Kinsey, S., Crowe, M., Ghosh, S., psychopharmacology 38, 2515e2524. https://doi.org/10.1038/npp.2013.157.
Owens, R.A., Damaj, I.M., Poklis, J., Wiley, J.L., Zanda, M., Zanato, C., Greig, I.R., Ortar, G., Cascio, M.G., De Petrocellis, L., Morera, E., Rossi, F., Schiano-Moriello, A.,
Lichtman, A.H., Ross, R.A., 2015. A cannabinoid CB1 receptor-positive allosteric Nalli, M., de Novellis, V., Woodward, D.F., Maione, S., Di Marzo, V., 2007. New N-
modulator reduces neuropathic pain in the mouse with No psychoactive effects. arachidonoylserotonin analogues with potential “dual” mechanism of action
Neuropsychopharmacology 40, 2948e2959. https://doi.org/10.1038/ against pain. J. Med. Chem. 50, 6554e6569. https://doi.org/10.1021/jm070678q.
npp.2015.148. Panlilio, L.V., Justinova, Z., Mascia, P., Pistis, M., Luchicchi, A., Lecca, S., Barnes, C.,
Ignatowska-Jankowska, B.M., Muldoon, P.P., Lichtman, A.H., Damaj, M.I., 2013. The Redhi, G.H., Adair, J., Heishman, S.J., Yasar, S., Aliczki, M., Haller, J., Goldberg, S.R.,
cannabinoid CB2 receptor is necessary for nicotine-conditioned place prefer- 2012. Novel use of a lipid-lowering fibrate medication to prevent nicotine
ence, but not other behavioral effects of nicotine in mice. Psychopharmacology reward and relapse: preclinical findings. Neuropsychopharmacology 37,
(Berl) 229, 591e601. https://doi.org/10.1007/s00213-013-3117-6. 1838e1847. https://doi.org/10.1038/npp.2012.31.
Jackson, A., Bagdas, D., Muldoon, P.P., Lichtman, A.H., Carroll, F.I., Greenwald, M., Perkins, K.A., Karelitz, J.L., Michael, V.C., Fromuth, M., Conklin, C.A.,
Miles, M.F., Damaj, M.I., 2017. In vivo interactions between a7 nicotinic Chengappa, K.N.R., Hope, C., Lerman, C., 2016. Initial evaluation of fenofibrate
acetylcholine receptor and nuclear peroxisome proliferator-activated receptor- for efficacy in aiding smoking abstinence. Nicotine Tob. Res. 18, 74e78. https://
a: implication for nicotine dependence. Neuropharmacology 118, 38e45. doi.org/10.1093/ntr/ntv085.
https://doi.org/10.1016/j.neuropharm.2017.03.005. Piscitelli, F., Carta, G., Bisogno, T., Murru, E., Cordeddu, L., Berge, K., Tandy, S.,
Jackson, K.J., Kota, D.H., Martin, B.R., Damaj, M.I., 2009. The role of various nicotinic Cohn, J.S., Griinari, M., Banni, S., Di Marzo, V., 2011. Effect of dietary krill oil
receptor subunits and factors influencing nicotine conditioned place aversion. supplementation on the endocannabinoidome of metabolically relevant tissues
Neuropharmacology 56, 970e974. https://doi.org/10.1016/ from high-fat-fed mice. Nutr. Metab. (Lond) 8, 51. https://doi.org/10.1186/1743-
j.neuropharm.2009.01.023. 7075-8-51.
G. Donvito et al. / Neuropharmacology 148 (2019) 320e331 331

Pushparaj, A., Hamani, C., Yu, W., Shin, D.S., Kang, B., Nobrega, J.N., Le Foll, B., 2013. Sugiura, T., Kondo, S., Sukagawa, A., Nakane, S., Shinoda, A., Itoh, K., Yamashita, A.,
Electrical stimulation of the insular region attenuates nicotine-taking and Waku, K., 1995. 2-Arachidonoylgylcerol: a possible endogenous cannabinoid
nicotine-seeking behaviors. Neuropsychopharmacology 38, 690e698. https:// receptor ligand in brain. Biochem. Biophys. Res. Commun 215, 89e97. https://
doi.org/10.1038/npp.2012.235. doi.org/10.1006/bbrc.1995.2437.
Pushparaj, A., Kim, A.S., Musiol, M., Trigo, J.M., Le Foll, B., 2015. Involvement of the Valjent, E., Mitchell, J.M., Besson, M.-J., Caboche, J., Maldonado, R., 2002. Behav-
rostral agranular insular cortex in nicotine self-administration in rats. Behav. ioural and biochemical evidence for interactions between D9-
Brain Res. 290, 77e83. https://doi.org/10.1016/j.bbr.2015.04.039. tetrahydrocannabinol and nicotine. Br. J. Pharmacol. 135, 564e578. https://
Robinson, J.D., Cinciripini, P.M., Karam-Hage, M., Aubin, H.-J., Dale, L.C., Niaura, R., doi.org/10.1038/sj.bjp.0704479.
Anthenelli, R.M., STRATUS Group, 2017. Pooled analysis of three randomized, Wang, J., Wolf, R.M., Caldwell, J.W., Kollman, P.A., Case, D.A., 2004. Development and
double-blind, placebo controlled trials with rimonabant for smoking cessation. testing of a general amber force field. J. Comput. Chem. 25, 1157e1174. https://
Addiction Biol. https://doi.org/10.1111/adb.12508. doi.org/10.1002/jcc.20035.
Ryckaert, J.-P., Ciccotti, G., Berendsen, H.J., 1977. Numerical integration of the car- Wang, S., Xu, Q., Shu, G., Wang, L., Gao, P., Xi, Q., Zhang, Y., Jiang, Q., Zhu, X., 2015. N-
tesian equations of motion of a system with constraints: molecular dynamics of Oleoyl glycine, a lipoamino acid, stimulates adipogenesis associated with acti-
n-alkanes. J. Comput. Phys. 23, 327e341. https://doi.org/10.1016/0021-9991(77) vation of CB1 receptor and Akt signaling pathway in 3T3-L1 adipocyte. Bio-
90098-5. chem. Biophys. Res. Commun. 466, 438e443. https://doi.org/10.1016/
Scheggi, S., Melis, M., De Felice, M., Aroni, S., Muntoni, A.L., Pelliccia, T., j.bbrc.2015.09.046.
Gambarana, C., De Montis, M.G., Pistis, M., 2016. PPARa modulation of meso- Watkins, S.S., Koob, G.F., Markou, A., 2000. Neural mechanisms underlying nicotine
limbic dopamine transmission rescues depression-related behaviors. Neuro- addiction: acute positive reinforcement and withdrawal. Nicotine Tob. Res. 2,
pharmacology 110, 251e259. https://doi.org/10.1016/j.neuropharm.2016.07.024. 19e37.
Simonnet, A., Cador, M., Caille, S., 2013. Nicotine reinforcement is reduced by Wiley, J.L., Martin, B.R., 2003. Cannabinoid pharmacological properties common to
cannabinoid CB1 receptor blockade in the ventral tegmental area. Addiction other centrally acting drugs. Eur. J. Pharmacol. 471, 185e193. https://doi.org/
Biol. 18, 930e936. https://doi.org/10.1111/j.1369-1600.2012.00476.x. 10.1016/S0014-2999(03)01856-9.

You might also like